WO2018081726A2 - Compositions pharmaceutiques et procédés d'utilisation pour l'activation de fibroblastes humains et d'apoptose de myofibroblastes - Google Patents

Compositions pharmaceutiques et procédés d'utilisation pour l'activation de fibroblastes humains et d'apoptose de myofibroblastes Download PDF

Info

Publication number
WO2018081726A2
WO2018081726A2 PCT/US2017/059072 US2017059072W WO2018081726A2 WO 2018081726 A2 WO2018081726 A2 WO 2018081726A2 US 2017059072 W US2017059072 W US 2017059072W WO 2018081726 A2 WO2018081726 A2 WO 2018081726A2
Authority
WO
WIPO (PCT)
Prior art keywords
tissue
composition
histidine
lysine
mammal
Prior art date
Application number
PCT/US2017/059072
Other languages
English (en)
Other versions
WO2018081726A3 (fr
Inventor
Jia Zhou
Qingfeng Li
John Xu
Patrick Y. Lu
Vero SIMONENKO
Original Assignee
Sirnaomics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sirnaomics, Inc. filed Critical Sirnaomics, Inc.
Priority to CN201780065567.9A priority Critical patent/CN110191712A/zh
Priority to US16/343,309 priority patent/US11697813B2/en
Priority to EP17865833.2A priority patent/EP3532071A4/fr
Publication of WO2018081726A2 publication Critical patent/WO2018081726A2/fr
Publication of WO2018081726A3 publication Critical patent/WO2018081726A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/495Transforming growth factor [TGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1136Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against growth factors, growth regulators, cytokines, lymphokines or hormones
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/0004Oxidoreductases (1.)
    • C12N9/0071Oxidoreductases (1.) acting on paired donors with incorporation of molecular oxygen (1.14)
    • C12N9/0083Miscellaneous (1.14.99)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2320/00Applications; Uses
    • C12N2320/30Special therapeutic applications

Definitions

  • the current invention relates to pharmaceutical compositions and methods for the activation of human fibroblast and myofibroblast apoptosis
  • Fibrosis is defined by excessive accumulation of extracellular matrix (ECM) in and around the damaged tissue, which can lead to permanent scarring (Miller et al, 2005).
  • ECM extracellular matrix
  • HTS Hypertrophic scar
  • Fibroblasts are the most common cells in connective tissue, playing a key role in the wound healing process and can differentiate into myofibroblasts that results in increased ECM synthesis and tissue contraction (McDougall et al, 2006 and Nedelec et al, 2001).
  • FIG. 1 Two potent siRNA duplexes targeting TGF- ⁇ and COX-2 respectively were selected using qRT-PCR analyses following cell transfections with corresponding siRNA dupelxes and total RNA solation.
  • the selected siRNA duplexes targeting TGF- ⁇ 1 and COX-2 exhibited high homology to their corresponding gene sequences of human, mouse, monkey and pig.
  • FIG. 2 Comparisons of target gene (TGF- ⁇ , COX-2, ⁇ - SMA, CollAl and Col3Al) mRNA expressions in human fibroblast cells after transfections with the TGF- ⁇ 1 siRNA , or COX-2 siRNA , or TGF- ⁇ 1/COX-2 siRNA s at 5 ⁇ g/ml (* ⁇ 0.05, ** p ⁇ 0.01). The total RNA samples were collected for each treated cell culture plate followed with qRT-PCR analysis. The results were demonstrated in the figure with error bars and statistic significant indicators. Where NS represents non-specific siRNA treatment control.
  • FIG. 3 Electron microscope images (SEM) of the fibroblast cells transfected with the TGF- ⁇ 1/COX-2 siRNA s illustrates an strong apoptotic activity, where N indicates nucleus, black arrows indicate Lipid-siRNA particles and red arrows indicate apoptosis bodies.
  • FIG. 4 FACS analysis indicates significant increase of apoptotic cell population of human myofibroblasts when TGF- ⁇ 1/COX-2 siRNAS was transfected simultaneously, as seen in the right lower panel, in comparison with the untreated and treated with the individual siRNA.
  • FIG. 4 Four different stages of treated fibroblasts: dying cells, apoptotic cells, viable cells and viable apoptotic cells. The cells treated with TGF- ⁇ 1/COX-2 siRNA in
  • FIG. 1 (Left) SEM image of FDCP (siRNA) nanoparticles were in a lyophilized form for long time storage and easy transportation. The nanoparticles were analyzed with a particle sizer (Brookhaven 190, NY, USA) and resulted in an average size of 150 nm ⁇ 30 in diameter.
  • (Right) SEM image of HKP (TGF- ⁇ 1/COX-2 siRNA s) nanoparticles in aqueous solution exhibited an average size of 150nm in diameter for injectable administration.
  • FIG. 7 List of physicochemical properties of HKP (siRNA) nanoparticles have indicated that the particle sizes (upper panel and lower left) and zeta-potentials of FDCP (siRNA) nanoparticles (middle panel and lower right) were measured, resulted in an average particles size about 150 nm in diameter, with Zeta-potentials of 38 vM.
  • Figure 8 Comparison between Alexa Fluor labeled siRNA and FDCP-packaged the same siRNA in vivo for their duration and dispersion after intra-scar injection. Samples collected from a human hypertrophic scar tissue implant model of mice, at 0, 24 and 48 hour time points post administrations.
  • FIG. 10 Injections of FDCP (TGF431/COX-2 siRNA s) nanoparticle solution into the human hypertrophic scar resulted in down regulations of TGF- ⁇ and COX-2 expressions in the tissue. This target gene silencing activity lasts up to 5 days, based on the qRT-PCR analyses following the one-shot administration.
  • FIG. 12 The process for establishing a human hypertrophic scar implant mouse model.
  • Key improvement to the original procedures for establishing such a model was that several stitches were performed to fix transplant to the skin pocket.
  • the human HTS transplant became very much viable when dissected one week later as seen with enrich blood vessel network.
  • Appearance images after the human hypertrophic scar implanted provided an overview of appearances after the implanted tissue from day 1, one-week, two-week, three- week, four-week, one-month and all the way to six -month.
  • FIG. 14 A close view of human skin implant on nude mouse shows both top and side views of a human skin sample implanted onto the nude mouse back. The tissue samples were collected and stained with H&E showing the implanted tissue has grown into the surrounding tissues.
  • Figure 15 The overview of the appearances of human skin tissue implanted onto nude mouse back, from two weeks to six months after the implantation. The observations was started from 2 weeks to 1 month, 2 month, 3 month, and all the way to 6 month.
  • Figure 18 Images of human skin implants, either treated with HKP (TGF431/COX- 2 siRNA s) or control aqueous solution, at day 0 and day 28 th post treatments. (Right)
  • FIG. 21 Tissue samples with H&E and Masson' s tri chrome staining, and IHC staining with antibodies against human VEGF, CD31 and ⁇ - SMA proteins, revealed down regulations of the angiogenesis, micro blood vessel marker and fibrogenesis after repeated treatments with HKP (TGF- ⁇ 1/COX-2siRNAs). Red arrows indicate epidermis layer of the skin.
  • HKP TGF- ⁇ 1/COX-2 siRNA s
  • fibroblast and myofibroblast apoptosis black arrows
  • Figure 23 Schematic demonstration of a novel mechanism for prevention and reduction of skin fibrotic scarring.
  • a normal skin wound healing process includes three phases:
  • STP705 treatment results in down regulation of TGF- ⁇ and COX-2 expressions and a fine-tuned fibroblast proliferation, which maintains a balance between deposition and degradation of ECM and promotes a scar- free wound healing.
  • Scar Reduction An optimal dosing of STP705 is expected to down regulate TGF- ⁇ 1 and COX-2 expressions in the scar tissue, resulting in apoptosis activation of fibroblasts/myofibroblasts within the scar tissue. The therapeutic effect of STP705 is to reverse fibrotic scarring and reduce hypertrophic scar.
  • the current invention provides a composition comprising an siRNA molecule that binds to an mRNA that codes for TGFBl protein in a mammalian cell, an siRNA molecule that binds to an mRNA that codes for COX-2 protein in a mammalian cell, and a
  • the current invention also provides methods of using the composition. In one embodiment, it provides a method of down-regulating pro-fibrotic factors and fibrotic pathways in the cells of a tissue of a mammal, comprising administering to the tissue a therapeutically effective amount of the composition. In another embodiment, it provides a method of activating fibroblast and myofibroblast apoptosis in a tissue of a mammal, comprising administering to the tissue a therapeutically effective amount of the composition.
  • the invention provides a method of reducing the size of a hypertrophic scar in the tissue of a mammal, comprising administering to the scar a therapeutically effective amount of the composition.
  • the invention provides a method of reducing fibrosis in the tissue of a mammal, comprising administering to the tissue a therapeutically effective amount of the composition.
  • the siRNA molecules can produce additive or synergistic effects in the cells, depending on the compositions and structures of the particular molecules. In a preferred embodiment, they produce a synergistic effect.
  • RNA molecule is a duplex oligonucleotide, that is a short, double-stranded polynucleotide, that interferes with the expression of a gene in a cell that produces RNA, after the molecule is introduced into the cell. For example, it targets and binds to a complementary nucleotide sequence in a single stranded (ss) target RNA molecule, such as an mRNA or a micro RNA (miRNA). The target RNA is then degraded by the cell.
  • ss target RNA molecule such as an mRNA or a micro RNA (miRNA).
  • the siRNA molecule can be made of naturally occurring ribonucleotides, i.e., those found in living cells, or one or more of its nucleotides can be chemically modified by techniques known in the art. In addition to being modified at the level of one or more of its individual nucleotides, the backbone of the oligonucleotide can be modified. Additional modifications include the use of small molecules (e.g. sugar molecules), amino acid molecules, peptides, cholesterol, and other large molecules for conjugation onto the siRNA molecule.
  • the molecule is an oligonucleotide with a length of about 19 to about 35 base pairs. In one aspect of this embodiment, the molecule is an oligonucleotide with a length of about 19 to about 27 base pairs. In another aspect, the molecule is an oligonucleotide with a length of about 21 to about 25 base pairs. In all of these aspects, the molecule may have blunt ends at both ends, or sticky ends at both ends, or a blunt end at one end and a sticky end at the other.
  • the relative amounts of the two different molecules and the copolymer can vary.
  • the ratio of the two different siRNA molecules is about 1 : 1 by mass.
  • the ratio of these molecules to the copolymer is about 1 :4, 1 :4.5, or 1 :5 by mass.
  • the ratio of the two different siRNA molecules is about 1 : 1 by mass and the ratio of these molecules to the copolymer is about 1 :4, 1 :4.5, or 1 :5 by mass.
  • the composition forms nanoparticles with an average size of about 150 nm in diameter.
  • the siRNA molecules are selected from the ones identified in Table 1.
  • An example is the pair designated hmTF-25-2 and hmCX-25-1 in the table, which has the following sequences:
  • hmTF-25-2 sense, 5'-r(CCCAAGGGCUACCAUGCCAACUUCU)-3',
  • hmCX-25-1 sense, 5'-r(GGUCUGGUGCCUGGUCUGAUGAUGU)-3 ,
  • the invention includes a method for identifying the desired siRNA molecules comprising the steps of: (a) creating a collection of siRNA molecules designed to target a complementary nucleotide sequence in the target mRNA molecules, wherein the targeting strands of the siRNA molecules comprise various sequences of nucleotides; (b) selecting the siRNA molecules that show the highest desired effect against the target mRNA molecules in vitro; (c) evaluating the selected siRNA molecules in an animal wound model; and (d) selecting the siRNA molecules that show the greatest efficacy in the model for their silencing activity and therapeutic effect.
  • siRNA sequences potentially targeting an mRNA sequence of a disease gene will, in fact, exhibit effective RNAi activity.
  • individually specific candidate siRNA polynucleotide or oligonucleotide sequences must be generated and tested in mammalian cell culture, such as an in vitro organ culture assay, to determine whether the intended interference with expression of a targeted gene has occurred.
  • the unique advantage of siRNA makes it possible to be combined with multiple siRNA duplexes to target multiple disease causing genes in the same treatment, since all siRNA duplexes are chemically homogenous with same source of origin and same manufacturing process.
  • a preferred animal wound model is a back skin excisional wound model in a Balb/c mouse or a back excisional wound model in a pig.
  • the animal wound model is a skin burn wound model in a pig.
  • the animal wound model is a back skin excisional wound model in a transgenic diabetic (db+/db+) mouse.
  • the siRNA molecules are evaluated in at least two of the animal models.
  • the method further includes the steps of adding a pharmaceutically acceptable carrier to each of the siRNA molecules selected by step (b) to form pharmaceutical compositions and evaluating each of the pharmaceutical compositions in the animal wound model or models.
  • the siRNA sequences are prepared in such way that each one can target and inhibit the same gene from, at least, both human and mouse, or human and non- human primate.
  • the siRNA molecules bind to both a human mRNA molecule and a homologous mouse mRNA molecule. That is, the human and mouse mRNA molecules encode proteins that are substantially the same in structure or function. Therefore, the efficacy and toxicity reactions observed in the mouse disease models provide a good understanding about what is going to happen in humans. More importantly, the siRNA molecules tested in the mouse model are good candidates for human pharmaceutical agents.
  • the human/mouse homology design of an siRNA drug agent can eliminate the toxicity and adverse effect of those species specificities observed in monoclonal antibody drugs.
  • the invention provides a composition comprising two or more different siRNA molecules that bind to an mRNA that codes for TGF ⁇ 1 protein in a mammalian cell and two or more different siRNA molecules that bind to an mRNA that codes for COX-2 protein in a mammalian cell.
  • the molecules may bind to different nucleotide sequences within the target mRNA.
  • the siRNA molecules can produce additive or synergistic effects in the cells, depending on the compositions and structures of the particular molecules. In a preferred embodiment, they produce a synergistic effect. In certain applications of these embodiments, the siRNA molecules are selected from the ones identified in Table 1.
  • the siRNA molecules are combined with a pharmaceutically acceptable carrier to provide pharmaceutical compositions for administering to a mammal.
  • the mammal is a laboratory animal, which includes dogs, cats, pigs, non-human primates, and rodents, such as mice, rats, and guinea pigs.
  • the mammal is a human.
  • the carrier is a histidine-lysine copolymer that forms a nanoparticle containing an siRNA molecule, wherein the nanoparticle has a size of 100-400 nm in diameter.
  • the carrier is selected from the group consisting of the HKP species, H3K4b and PT73, which have a Lysine backbone with four branches containing multiple repeats of Histidine, Lysine, or Asparagine.
  • the HKP has the following formula:
  • the HKP has the following formula:
  • compositions of the invention are useful for down-regulating pro-fibrotic factors, such as ⁇ - smooth muscle actin ( ⁇ - SMA), Hydroxyproline Acid, Smad 3, and Connective Tissue Growth Factor (CTGF), and fibrotic pathways, such as TGF- ⁇ 1/Smad 3/ ⁇ - SMA/Collagen I-III, in the cells of a tissue of a mammal.
  • pro-fibrotic factors such as ⁇ - smooth muscle actin ( ⁇ - SMA), Hydroxyproline Acid, Smad 3, and Connective Tissue Growth Factor (CTGF), and fibrotic pathways, such as TGF- ⁇ 1/Smad 3/ ⁇ - SMA/Collagen I-III
  • fibrotic pathways such as TGF- ⁇ 1/Smad 3/ ⁇ - SMA/Collagen I-III
  • the tissue is skin scar, liver, lung, kidney, or heart tissue.
  • the tissue is skin scar tissue.
  • the cells comprise fibroblasts and myofibroblasts.
  • the fibroblasts and myofibroblasts are dermal fibroblasts and myofibroblasts.
  • the mammal is a human.
  • compositions of the invention are also useful for activating fibroblast and myofibroblast apoptosis in the tissue of a mammal. This reduces tissue fibrosis caused by scarring after chronic inflammation of the tissue.
  • a therapeutically effective amount of the composition is administered to the tissue of the mammal.
  • Such apoptosis may be determined and measured by measuring the apoptotic cell population with FACS analysis, counting body numbers, and detecting expression levels of TGF- ⁇ , COX-2, a-SMA, Collagen I and
  • the tissue is skin scar, liver, lung, kidney, or heart tissue. In one aspect of this embodiment, the tissue is skin scar tissue. In another embodiment, the fibroblasts and myofibroblasts are dermal fibroblasts and myofibroblasts. Preferably, the mammal is a human.
  • One particular embodiment of the invention provides a method of activating fibroblast and myofibroblast apoptosis in a tissue of a human, comprising injecting into the tissue a therapeutically effective amount of a composition comprising the siRNA molecule hmTF-25- 2: sense, 5'-r(CCCAAGGGCUACCAUGCCAACUUCU)-3 ', antisense, 5 '- r(AGAAGUUGGCAUGGUAGCCCUUGGG)-3 ' , the siRNA molecule hmCX-25-1 : sense, 5'-r(GGUCUGGUGCCUGGUCUGAUGAUGU)-3 ', antisense, 5'- r(ACAUCAUCAGACCAGGCACCAGACC)-3 ⁇ and a pharmaceutically acceptable carrier comprising a pharmaceutically acceptable histidine-lysine co-polymer.
  • the histidine-lysine co-polymer comprises the histidine-lysine co-polymer species H3K4b or the histidine-lysine co-polymer species PT73.
  • compositions of the invention are also useful for reducing the size of a hypertrophic scar in the tissue of a mammal.
  • a therapeutically effective amount of the composition is administered to the scar tissue.
  • tissue includes, but is not limited to, skin, liver, lung, kidney, and heart tissue.
  • the scar comprises fibroblasts and myofibroblasts.
  • the scar comprises dermal fibroblasts in dermal myofibroblasts.
  • the mammal may be a laboratory animal, such as a dog, cat, pig, non-human primate, or rodent, such as a mouse, rat, or guinea pig.
  • the mammal is a human.
  • hypertrophic scar formation is reversed.
  • One particular embodiment of the invention provides a method of reducing the size of a hypertrophic scar in the skin tissue of a human, comprising injecting into the scar tissue a therapeutically effective amount of a composition comprising the siRNA molecule hmTF-25- 2: sense, 5'-r(CCCAAGGGCUACCAUGCCAACUUCU)-3 ', antisense, 5 '- r(AGAAGUUGGCAUGGUAGCCCUUGGG)-3 ' , the siRNA molecule hmCX-25-1 : sense, 5'-r(GGUCUGGUGCCUGGUCUGAUGAUGU)-3 ', antisense, 5'- r(ACAUCAUCAGACCAGGCACCAGACC)-3 ⁇ and a pharmaceutically acceptable carrier comprising a pharmaceutically acceptable histidine-lysine co-polymer.
  • the histidine-lysine co-polymer comprises the histidine-lysine co-polymer species H3K4b or the histidine-lysine co-polymer species PT73.
  • compositions of the invention are also useful for reducing fibrosis in the tissue of a mammal.
  • a therapeutically effective amount of the composition is delivered to the tissue.
  • tissue includes, but is not limited to, skin, liver, lung, kidney, and heart tissue.
  • the fibrotic tissue comprises fibroblasts and myofibroblasts.
  • the composition may be delivered by injection into the tissue, subcutaneous injection into the mammal, or intravenous injection into the mammal.
  • the mammal may be a laboratory animal, such as a dog, cat, pig, non-human primate, or rodent, such as a mouse, rat, or guinea pig.
  • the mammal is a human.
  • the composition is administered by injection into the tissue.
  • the composition is ministered by subcutaneous injection into the mammal.
  • the composition is administered intravenously to the mammal.
  • the mammal is a human.
  • siRNA sequences specific to human TGF- ⁇ ⁇ and COX-2 mRNAs were based on efficient cell transfection studies and analysis using qRT-PCR ( Figure 1).
  • the potent siRNAs we selected were based on their silencing efficiencies and toxicity profiles affecting cells.
  • human fibroblasts isolated from the hypertrophic scar tissue were transfected with the selected siRNAs targeting either TGF- ⁇ or COX-2 individually or in combination, we observed an efficient siRNA entry into the cells at different stages, from initial endocytosis to endosome release of the siRNAs.
  • Example 2 Simultaneous silencing of TGF- ⁇ and COX-2 gene expressions resulted in activation of apoptosis of fibroblasts/myofibroblasts
  • HPC hydroxyproline acid
  • Example 3 HKP enhances siRNA delivery into human hypertrophic scar
  • HKP biodegradable histidine-lysine polypeptides
  • the lyophilized HKP (TGF- ⁇ 1 /COX-2siRNA) nanoplex powder with an average size about 150 nm in diameter and a Zeta potential about 40 mV, is highly stable at 4°C, preserving potent activity for silencing TGF- ⁇ expression up to 12 months (Figure 7).
  • TGF- ⁇ 1 and COX-2 are significantly over-expressed in the human hypertrophic scar (HTS) tissue from patients scar biopsy compared to the normal skin tissue (Figure 10).
  • HTS human hypertrophic scar
  • Figure 10 Human HTS tissues were implanted onto nude mice subcutaneously for studying pathophysiology of HTS and its novel therapeutic options. After HTS tissue was implanted, we collected some of these tissues at day 7, day 14 and day 28 post implantation. We then isolated mRNA from those tissue samples for evaluating the expression dynamics of TGF- ⁇ ⁇ , COX-2 and ⁇ - SMA using qRT-PCR analyses (Figure 11).
  • TGF- ⁇ and COX-2 in the implanted human scar tissues exhibited a unique pattern with immediate escalation of TGF- ⁇ versus a steady increase in COX-2.
  • the TGF- ⁇ expression reached a peak level at day 14 while COX-2 expression was still up regulated on day 28 post initial implantation.
  • HKP TGF- ⁇ 1 /COX-2 siRNAs reduces size of human skin grafts
  • human skin grafted onto the nude mouse is able to regenerate after being subjected to a full-thickness wound.
  • This approach has been used to determine the cells involved in the connective tissue repair process following superficial wounding.
  • this model has been used to study the wound healing process of human skin.
  • the hypertrophic scar model is established by transplanting human skin grafts onto nude mice, resulting in obvious, persistent hypertrophic scars that have both macroscopic and histologic properties similar to human hypertrophic scars. This model makes possible the observation of the entire process of hypertrophic scar formation. Thus, it is an ideal tool for studying hypertrophic scar (Yang, et al. 2007).
  • the initial dosing time and dosing regimen were similar to the treatment of the implanted human hypertrophic scars on mice.
  • 20 ⁇ g/50 ⁇ l/cm 3 HKP (TGF- ⁇ 1/COX-2 SI RNA S ) solution was injected into each skin graft using 5 aliquots to 5 different sites of the graft, with three repeated injections at 5 day intervals.
  • the HKP (TGF- ⁇ 1 /COX-2 siRNA s) combination treated human skin grafts resulted in a significant size reduction at day 28 post-treatment (Figure 18), about 40% in comparison to the untreated group.
  • HKP TGF- ⁇ 1 /COX-2 siRNAs
  • Example 7 HKP (TGF- ⁇ 1 /COX-2 siRNAs) induced apoptotic activities of the fibroblast/myofibroblasts in vivo
  • HKP TGF- ⁇ 1 /COX-2 siRNAs
  • HKP TGF- ⁇ 1 /COX-2 siRNAs silencing both TGF- ⁇ and COX-2 at proliferation stage and remodeling stage of skin wound healing process
  • a dermal wound healing process can be specified into three phases: inflammation, cell proliferation and matrix remodeling, which involve multiple interactions within a complex network of pro-fibrotic and anti-fibrotic molecules (Dabiri et al, 2006).
  • the aggregated inflammatory cells become sources of growth factors and cytokines.
  • active angiogenesis and collagen synthesis ensue in concert with the tissue remodeling process, a delicate balance of deposition and degradation of fibroblast- expressed ECM determines normal skin wound healing or whether a wound heals but with HTS.
  • Fibroblasts are the most common cells in connective tissue and key players in skin wound healing process, functioning to maintain the physical integrity of the connective tissue, participate in wound closure, and produce and remodel ECM (Wang et al, 2011).
  • fibroblast apoptosis in the cell culture and human HTS and skin tissue implants confirmed a critical therapeutic potential of FDCP (TGF- ⁇ 1 /COX-2 SI RNA S ), in maintaining an optimized fibroblast proliferation and a balance between deposition and degradation of ECM production, to avoid fibrotic scarring.
  • FDCP TGF- ⁇ 1 /COX-2 SI RNA S
  • ⁇ - SMA, Collagen 1, Collagen 3 and hydroxproline acid in both human fibroblasts and HTS tissue at both mRNA and protein levels after the treatments, implicates a complex network regulating skin fibrotic scarring.
  • the results from this study have further advanced our understanding of the mechanism of actions of the pathophysiological pathways involved in the hypertrophic scar formation.
  • siRNA duplexes targeting TGF ⁇ 1 or COX-2 mRNA sequences were designed (Table 1). (Does this refer to Table 1; i.e., should "Supplemented” be deleted?) Eight siRNA for each gene were screened in human PC3 cells for target gene silencing with qRT-PCR analyses.
  • Cells were seeded into the wells of 96-well plate at density 2xl0 3 cells per well in lOOul media Six hours later culture media was replaced with fresh media supplemented containing Lipofectamine 2000 (Lipo2000) formulations, Lipo (TGF- ⁇ 1/COX-2 siRNA s), or Lipo2000, or Lipo2000 (TGF- ⁇ 1SIRNA) or Lipo2000 (COX-2 SIRN A). The cells were incubated for 48 hours. For growth inhibition assay, cells were treated and analyzed with target gene silencing, FACS, ⁇ - SMA and Hydroxylproline Acid expressions.
  • Lipofectamine 2000 Lipofectamine 2000
  • Lipo TGF- ⁇ 1/COX-2 siRNA s
  • Lipo2000 Lipo2000
  • TGF- ⁇ 1SIRNA Lipo2000
  • COX-2 SIRN A Lipo2000
  • HKP Histidine-Lysine polymers
  • mice 8-week old male nude mice (nu/nu Balb/c) were purchased from Center for Experimental Animals in Shanghai, China. Animal housing and experiment protocols were approved by the IACUC committee of the 9 th People's Hospital of Shanghai.
  • Skin hypertrophic scar tissue was obtained from the surgical excisions with the informed consent were implanted under the skin on the mouse back. Scar tissue was fixed to the mouse deep fascia with 4-5 sutures before cut on skin was closed.
  • the skin tissue samples used in experiments were from skin excisions from three women of age 23-36 undergone breast reconstruction for treatment of macromastia with signed informed consent. The skin tissues were grafted to fill the excision wounds by sutures to subcutaneous fascia and surrounding mouse skin. Therapeutic evaluation with the human tissue implant models
  • HKP TGF- ⁇ 1/COX-2 siRNA s
  • the mRNA levels of TGF- ⁇ , COX-2, a-SMA, Col lal, and Col3al were analyzed with qRT-PCR
  • the In Situ Cell Death Detection Kit from Roche was applied for detection of apoptotic cells.
  • Mean ⁇ SD was used for cell culture results, and mean ⁇ SE was used for in vivo results.
  • An unequal variance two-tailed Student's t test was applied to compare the means of samples. A difference was considered statistically significant when P ⁇ 0.05.
  • hypertrophic scar-like nude mouse model characterization of the molecular and cellular biology of the scar process. Wound Repair Regen. 2011. 19, 2:274-85.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Biotechnology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Endocrinology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Inorganic Chemistry (AREA)
  • Virology (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Dermatology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention concerne une méthode d'activation de fibroblastes et d'apoptose de myofibroblastes dans un tissu d'un mammifère, comprenant l'administration au tissu d'une quantité thérapeutiquement efficace d'une composition comprenant une petite molécule d'ARNi qui se lie à un ARNm codant pour la protéine TGFBl dans une cellule de mammifère, une petite molécule d'ARNi qui se lie à un ARNm codant pour la protéine COX-2 dans une cellule de mammifère, et un vecteur pharmaceutiquement acceptable comprenant un polymère d'histidine-lysine pharmaceutiquement acceptable. L'invention concerne également des méthodes d'utilisation supplémentaires de ladite composition.
PCT/US2017/059072 2016-10-30 2017-10-30 Compositions pharmaceutiques et procédés d'utilisation pour l'activation de fibroblastes humains et d'apoptose de myofibroblastes WO2018081726A2 (fr)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN201780065567.9A CN110191712A (zh) 2016-10-30 2017-10-30 用于激活人成纤维细胞和肌成纤维细胞凋亡的药物组合物及方法
US16/343,309 US11697813B2 (en) 2016-10-30 2017-10-30 Pharmaceutical compositions and methods of use for activation of human fibroblast and myofibroblast apoptosis
EP17865833.2A EP3532071A4 (fr) 2016-10-30 2017-10-30 Compositions pharmaceutiques et procédés d'utilisation pour l'activation de fibroblastes humains et d'apoptose de myofibroblastes

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662414780P 2016-10-30 2016-10-30
US62/414,780 2016-10-30

Publications (2)

Publication Number Publication Date
WO2018081726A2 true WO2018081726A2 (fr) 2018-05-03
WO2018081726A3 WO2018081726A3 (fr) 2018-05-31

Family

ID=62025520

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/059072 WO2018081726A2 (fr) 2016-10-30 2017-10-30 Compositions pharmaceutiques et procédés d'utilisation pour l'activation de fibroblastes humains et d'apoptose de myofibroblastes

Country Status (4)

Country Link
US (1) US11697813B2 (fr)
EP (1) EP3532071A4 (fr)
CN (1) CN110191712A (fr)
WO (1) WO2018081726A2 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10421970B2 (en) 2004-05-12 2019-09-24 Thermo Fisher Scientific Inc. Methods and compositions for selecting siRNA of improved functionality
WO2020034001A1 (fr) * 2018-08-14 2020-02-20 Loxegen Holdings Pty Ltd Nanoparticules pour la transfection
CN112703196A (zh) * 2018-05-24 2021-04-23 圣诺制药公司 用于核酸治疗的可控偶联多肽纳米颗粒导入系统的组合物及方法
CN114144423A (zh) * 2018-12-27 2022-03-04 圣诺制药公司 使用与免疫检查点抑制剂组合递送的siRNA沉默TGF-BETA 1和COX2以治疗癌症
CN115463148A (zh) * 2021-06-11 2022-12-13 圣诺生物医药技术(苏州)有限公司 一种用于治疗皮肤肿瘤的小干扰核酸药物组合物及制剂

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023049814A2 (fr) * 2021-09-22 2023-03-30 Sirnaomics, Inc. Compositions pharmaceutiques de nanoparticules présentant une taille de nanoparticules réduite et un indice de polydispersité amélioré
WO2023092142A1 (fr) * 2021-11-22 2023-05-25 Sirnaomics, Inc. Procédés pour induire le remodelage du tissu adipeux à l'aide de produits thérapeutiques à base d'arni

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101897982A (zh) * 2009-05-31 2010-12-01 苏州圣诺生物医药技术有限公司 治疗癌症的siRNA药物组合物
US9642873B2 (en) 2010-05-04 2017-05-09 Sirnaomics, Inc. Combinations of TGFβ and COX-2 inhibitors and methods for their therapeutic application
CN102031260A (zh) * 2010-08-24 2011-04-27 苏州圣诺生物医药技术有限公司 促进皮肤伤口无疤痕愈合的siRNA及应用
US20140072613A1 (en) * 2012-09-10 2014-03-13 Cynthia Lander Compositions and Methods for Treating Cutaneous Scarring
US20150065431A1 (en) * 2013-08-27 2015-03-05 Northwestern University Reducing cutaneous scar formation and treating skin conditions
CN103642800A (zh) 2013-11-19 2014-03-19 上海交通大学医学院附属第九人民医院 siRNA分子组合物及其在治疗增生性瘢痕中的用途
CN104174032A (zh) 2014-08-06 2014-12-03 上海交通大学医学院附属第九人民医院 siRNA分子组合物及其在治疗病理性瘢痕中的用途

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10421970B2 (en) 2004-05-12 2019-09-24 Thermo Fisher Scientific Inc. Methods and compositions for selecting siRNA of improved functionality
CN112703196A (zh) * 2018-05-24 2021-04-23 圣诺制药公司 用于核酸治疗的可控偶联多肽纳米颗粒导入系统的组合物及方法
JP2021525508A (ja) * 2018-05-24 2021-09-27 サーナオミクス インコーポレイテッド 核酸治療薬のための調節可能な共カップリングポリペプチドナノ粒子送達系の組成物および方法
EP3801025A4 (fr) * 2018-05-24 2022-03-09 Sirnaomics, Inc. Composition et procédés de système d'administration de nanoparticules polypeptidiques à co-couplage contrôlable pour des agents thérapeutiques à base d'acides nucléiques
WO2020034001A1 (fr) * 2018-08-14 2020-02-20 Loxegen Holdings Pty Ltd Nanoparticules pour la transfection
EP3836952A4 (fr) * 2018-08-14 2023-01-04 Loxegen Holdings Pty., Ltd. Nanoparticules pour la transfection
CN114144423A (zh) * 2018-12-27 2022-03-04 圣诺制药公司 使用与免疫检查点抑制剂组合递送的siRNA沉默TGF-BETA 1和COX2以治疗癌症
CN115463148A (zh) * 2021-06-11 2022-12-13 圣诺生物医药技术(苏州)有限公司 一种用于治疗皮肤肿瘤的小干扰核酸药物组合物及制剂

Also Published As

Publication number Publication date
CN110191712A (zh) 2019-08-30
EP3532071A4 (fr) 2020-09-02
EP3532071A2 (fr) 2019-09-04
US11697813B2 (en) 2023-07-11
WO2018081726A3 (fr) 2018-05-31
US20200392507A1 (en) 2020-12-17

Similar Documents

Publication Publication Date Title
US11697813B2 (en) Pharmaceutical compositions and methods of use for activation of human fibroblast and myofibroblast apoptosis
Lai et al. Tailored design of electrospun composite nanofibers with staged release of multiple angiogenic growth factors for chronic wound healing
Rodness et al. VEGF-loaded microsphere patch for local protein delivery to the ischemic heart
Gondaliya et al. Mesenchymal stem cell-derived exosomes loaded with miR-155 inhibitor ameliorate diabetic wound healing
Takahashi et al. Device-based local delivery of siRNA against mammalian target of rapamycin (mTOR) in a murine subcutaneous implant model to inhibit fibrous encapsulation
Castleberry et al. Nanolayered siRNA delivery platforms for local silencing of CTGF reduce cutaneous scar contraction in third-degree burns
Gan et al. Antibacterial, adhesive, and MSC exosomes encapsulated microneedles with spatio-temporal variation functions for diabetic wound healing
JP2015096078A (ja) 創傷治癒組成物および治療
JP6140120B2 (ja) 挫瘡およびその他の状態を治療するためのビスファチン治療薬
WO2011140285A2 (fr) Combinaisons d'inhibiteurs de tgfβ et de cox-2 et leurs méthodes d'utilisation thérapeutique
JP2011504170A (ja) 皮膚の中で平癒している瘢痕のない損傷のためのマルチ目標とされたRNAi治療学
JP2021506943A (ja) 乳房障害の治療の乳管内送達方法
JP2011507857A (ja) 線維症性の状態の処置のためのコネキシン43の阻害剤の使用
US20210340541A1 (en) Methods and compositions to inhibit metastasis and to treat fibrosis and to enhance wound healing
Yang et al. Exosomes from young healthy human plasma promote functional recovery from intracerebral hemorrhage via counteracting ferroptotic injury
EP3532088A1 (fr) Utilisation d'un sirna pour le traitement des cancers
CN107982536B (zh) 一种药物作用靶点的组合物和应用
CN110229215A (zh) 一种药物缓释多肽水凝胶及其制备方法和用途
Dankers et al. Convenient formulation and application of a supramolecular ureido-pyrimidinone modified poly (ethylene glycol) carrier for intrarenal growth factor delivery
KR20170096713A (ko) 이식용 줄기세포 및 이를 포함하는 혈관신생의 유도 또는 촉진을 위한 이식용 약학 조성물
CN111978405A (zh) 功能多肽、可特异结合胶原的红细胞载药体系及其应用
US20230302191A1 (en) Compositions and methods for sustained oxygen release to ischemic tissues
WO2018022457A1 (fr) Cicatrisation accélérée grâce à des supports amphiphiles ramifiés à plusieurs branches injectables chargés de médicaments hydrophobes
CN115837005A (zh) 一种传递体组合物及其制备方法和应用
CN114949245A (zh) 用于增强向皮肤角质形成细胞的局部递送和治疗皮肤炎症的自我治疗纳米颗粒

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 17865833

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2017865833

Country of ref document: EP

Effective date: 20190531